Attachment 1: Product information for AusPAR Juvicor/Xelezor/Tesozor Sitagliptin and Simvastatin, Merck, Sharp & Dohme Australia Pty Ltd PM-2011-02796-3-5. This Product Information was approved at the time this AusPAR was published.

Product information

JUVICOR®

(sitagliptin phosphate monohydrate/simvastatin, MSD)

JUVICOR 100/10 mg

JUVICOR 100/20 mg

JUVICOR 100/40 mg

Name of the medicine

JUVICORtablets contain sitagliptin (as phosphate monohydrate) and simvastatin.

Sitagliptin phosphate monohydrate (CAS no.: 654671-77-9), is described chemically as:
7-[(3R)-3-amino-1-oxo-4-(2,4,5-trifluorophenyl)butyl]-5,6,7,8-tetrahydro-3-(trifluoromethyl)-1,2,4-triazolo[4,3-a]pyrazine phosphate (1:1) monohydrate. The empirical formula is C16H15F6N5O•H3PO4•H2O and the molecular weight is 523.32. The structural formula is:

Simvastatin(CAS no.: 79902-63-9), is described chemically as
[1S-[1,3,7,8(2S*,4S*),8]]-1,2,3,7,8,8a-hexahydro-3,7-dimethyl-8-[2-(tetrahydro-4-hydroxy-6-oxo-2H-pyran-2-yl)ethyl]-1-naphthalenyl 2,2-dimethylbutanoate. The empirical formula is C25H38O5 and the molecular weight is 418.57. The structural formula is:

Description

Sitagliptin phosphate monohydrate is a white to off-white, crystalline, non-hygroscopic powder. It is soluble in water and N,N-dimethyl formamide; slightly soluble in methanol; very slightly soluble in ethanol, acetone, and acetonitrile; and insoluble in isopropanol and isopropyl acetate. The pH of a saturated water solution of sitagliptin phosphate monohydrate is 4.4. The partition coefficient is 1.8 and the pKa is 7.7. Simvastatin is a white crystalline powder, practically insoluble in water and freely soluble in chloroform, methanol and ethanol. The partition coefficient is >1995. Simvastatin exhibits no acid/base dissociation constants.

JUVICORis available for oral use as film-coated bilayer tablets containing128.5mg of sitagliptin phosphate monohydrate(equivalent to 100mg of free base), and either 10mg, 20mg, or 40mg of simvastatin. Each bilayer tablet of JUVICORalso contains the following inactive ingredients: anhydrous calcium hydrogen phosphate, microcrystalline cellulose, croscarmellose sodium, sodium stearylfumarate, magnesium stearate, ascorbic acid, citric acid monohydrate, lactose, and pregelatinised maizestarch. In addition, the film coating contains the following inactive ingredients: polyvinyl alcohol, macrogol3350, purified talc, titanium dioxide, iron oxide red CI77491, iron oxide yellow CI77492, and iron oxideblack CI77499. Butylated hydroxyanisole is added as an antioxidant.

Pharmacology

Pharmacodynamics

Sitagliptin

Sitagliptin phosphate monohydrate is an orally-active inhibitor of the dipeptidyl peptidase4 (DPP4) enzyme for the treatment of type 2 diabetes mellitus. The DPP4 inhibitors are a class of agents that act as incretin enhancers. By inhibiting the DPP4 enzyme, sitagliptin increases the levels of two known active incretin hormones, glucagon-like peptide1 (GLP1) and glucose-dependent insulinotropic polypeptide (GIP). The incretins are part of an endogenous system involved in the physiological regulation of glucose homeostasis. When blood glucose concentrations are normal or elevated, GLP1 and GIP increase insulin synthesis and release from pancreatic beta cells. GLP1 also lowers glucagon secretion from pancreatic alpha cells, leading to reduced hepatic glucose production. This mechanism is unlike the mechanism seen with sulfonylureas; sulfonylureas cause insulin release even when glucose levels are low, which can lead to sulfonylurea-induced hypoglycaemia in patients with type 2 diabetes and in normal subjects. Sitagliptin inhibits DPP4 with nanomolar potency (IC50 18 nM). It does not inhibit the closely-related enzymes DPP8 or DPP9 at therapeutic concentrations.

Sitagliptin differs in chemical structure and pharmacological action from GLP1 analogues, insulin, sulfonylureas or meglitinides, biguanides, peroxisome proliferator-activated receptor gamma (PPAR) agonists, alpha-glucosidase inhibitors, and amylin analogues.

In patients with type 2 diabetes, administration of single oral doses of sitagliptin leads to inhibition of DPP4 enzyme activity for a 24hour period, resulting in a 2 to 3fold increase in circulating levels of active GLP1 and GIP, increased plasma levels of insulin and C-peptide, decreased glucagon concentrations, reduced fasting glucose, and reduced glucose excursion following an oral glucose load or a meal. In a study of patients with type 2 diabetes inadequately controlled on metformin monotherapy, glucose levels monitored throughout the day were significantly lower in patients who received sitagliptin 100mg per day (50mg twice daily) in combination with metformin compared with patients who received placebo with metformin (see Figure1).

Figure1: 24hour Plasma Glucose Profile after 4Week Treatment with Sitagliptin 50mg BID with Metformin or Placebo with Metformin

In PhaseIII clinical studies of 18 and 24week duration, treatment with sitagliptin 100mg daily in patients with type 2 diabetes significantly improved beta cell function, as assessed by several markers, including HOMAβ (Homeostasis Model Assessmentβ), proinsulin to insulin ratio, and measures of beta cell responsiveness from the frequently-sampled meal tolerance test. There are no clinical studies that demonstrate that sitagliptin alters the natural history of impaired glucose tolerance or type 2 diabetes mellitus. The durability of efficacy requires further study.

In PhaseII studies, sitagliptin 50mg twice daily provided no additional glycaemic efficacy compared to 100mg once daily.

In studies with healthy subjects, sitagliptin did not lower blood glucose or cause hypoglycaemia, suggesting that the insulinotropic and glucagon suppressive actions of sitagliptin are glucose dependent (see PRECAUTIONS, Hypoglycaemia in Combination with a Sulfonylurea or with insulin;ADVERSE EFFECTS).

Effects on blood pressure

In a randomised, placebo-controlled crossover study in hypertensive patients on one or more anti-hypertensive medicines (including angiotensin-converting enzyme [ACE] inhibitors, angiotensinII antagonists, calcium-channel blockers, beta-blockers and diuretics), co-administration with sitagliptin was generally well tolerated. In these patients, sitagliptin had a modest blood pressure lowering effect; 100mg per day of sitagliptin reduced 24hour mean ambulatory systolic blood pressure by approximately 2mmHg, as compared to placebo. Reductions have not been observed in subjects with normal blood pressure.

Cardiac electrophysiology

In a randomised, placebo-controlled crossover study, 79healthy subjects were administered a single oral dose of sitagliptin 100mg, sitagliptin 800mg (8times the recommended dose), and placebo. At the recommended dose of 100mg, there was no effect on the QTc interval obtained at the peak plasma concentration, or at any other time during the study. Following the 800mg dose, the maximum increase in the placebo-corrected mean change in QTc from baseline at 3hours post-dose was 8.0msec. This small increase was not considered to be clinically significant. At the 800mg dose, peak sitagliptin plasma concentrations were approximately 11times higher than the peak concentrations following a 100mg dose. In patients with type 2 diabetes administered sitagliptin 100mg (N=81) or sitagliptin 200mg (N=63) daily, there were no meaningful changes in QTc interval based on ECG data obtained at the time of expected peak plasma concentration.

Simvastatin

Simvastatin is a lipid-lowering agent derived synthetically from a fermentation product of Aspergillus terreus. After oral ingestion, simvastatin, which is an inactive lactone, is hydrolysed to the corresponding-hydroxyacid form. This is a principal metabolite and an inhibitor of 3-hydroxy-3-methylglutaryl-coenzyme A (HMGCoA) reductase, an enzyme which catalyses an early and rate-limiting step in the biosynthesisof cholesterol. As a result, in clinical studies, simvastatin reduced total plasma cholesterol (totalC), low-density lipoprotein cholesterol (LDLC), and very-low-density lipoprotein cholesterol (VLDLC) concentrations. In addition, simvastatin increases high-density lipoprotein cholesterol (HDLC) and reduces plasma triglycerides (TG).

Simvastatin has been shown to reduce both normal and elevated LDLC concentrations. LDL is formed from VLDL and is catabolised predominantly by the high affinity LDL receptor. The mechanism of the LDLlowering effect of simvastatin may involve both reduction of VLDLC concentration and induction of the LDL receptor, leading to reduced production and increased catabolism of LDLC. Apolipoprotein B (Apo B) also falls substantially during treatment with simvastatin. Since each LDL particle contains one molecule of Apo B, and since little Apo B is found in other lipoproteins, this strongly suggests that simvastatin does not merely cause cholesterol to be lost from LDL, but also reduces the concentration of circulating LDL particles. As a result of these changes, the ratios of total-C to HDLC and LDL-C to HDLC are reduced.

Even though simvastatin is a specific inhibitor of HMGCoA reductase, the enzyme which catalyses the conversion of HMGCoA to mevalonate is not completely blocked at therapeutic doses, thereforeit allows the necessary amounts of mevalonate to be available for biological functions. Because the conversion of HMGCoA to mevalonate is an early step in the biosynthetic pathway of cholesterol, therapy with simvastatin would not be expected to cause an accumulation of potentially toxic sterols. In addition, HMGCoA is metabolised readily back to acetylCoA, which participates in many biosynthetic processes in the body.

Epidemiological studies have demonstrated that elevated levels of totalC, LDLC, as well as decreased levels of HDLC are associated with the development of atherosclerosis and increased cardiovascular risk. Lowering LDLC decreases this risk. However, the independent effect of raising HDLC or lowering TG on the risk of coronary and cardiovascular morbidity and mortality has not been determined.

Pharmacokinetics

Absorption

An increase in simvastatin acid mean peak plasma concentration (Cmax) was observed following co-administration of JUVICOR with a high-fat meal. The pharmacokinetics of sitagliptin were not affected under the same conditions. However, relative to the fasting state, the plasma profile of HMG-CoA reductase inhibitors was not affected when simvastatin was administered immediately before a test (i.e., non-high-fat) meal. JUVICORmay therefore be administered with or without food, however co-administration with a high-fat meal is not recommended.

The pharmacokinetics of sitagliptinhave been extensively characterised in healthy subjects and patients with type 2 diabetes. After oral administration of a 100mg dose to healthy subjects, sitagliptin was rapidly absorbed, with peak plasma concentrations (median Tmax) occurring 1 to 4hours post-dose. Plasma area under the curve (AUC) of sitagliptin increased in a dose-proportional manner. Following a single oral 100mg dose to healthy volunteers, mean plasma AUC of sitagliptin was 8.52M•hr, mean plasmaCmax was 950nM, and apparent terminal halflife (t1/2) was 12.4hours. Plasma AUC of sitagliptin increased approximately 14% following 100mg doses at steady-state compared to the first dose. The intra-subject and inter-subject coefficients of variation for sitagliptin AUC were small (5.8% and 15.1%). The pharmacokinetics of sitagliptin was generally similar in healthy subjects and in patients with type 2 diabetes. The absolute bioavailability of sitagliptin is approximately 87%.

Peak plasma concentrations of simvastatin and β-hydroxyacid were attained within 1.5 and 4-6hours post-dose, respectively. Based on assays of HMGCoA inhibition, no substantial deviation from linearity of AUC of inhibitors in the general circulation was observed at doses up to 120mg. The pharmacokinetic effects of calcium channel blockers on simvastatin and HMG-CoA reductase inhibitors are summarised in Table 1. The data show increases in simvastatin acid exposure (AUC) with calcium channel blockers (see PRECAUTIONS, Myopathy/Rhabdomyolysis).

Table 1 Effect of Co-administered Calcium Channel Blockers on Simvastatin Systemic Exposure and HMGCoA Reductase Inhibitory Activity

Geometric mean ratio (Ratio* with / without co-administered medicine) No Effect = 1.00
Co-administered medicine and dosing regimen / Dosing of Simvastatin / AUC / Cmax
Verapamil SR
240mg QD Days 1-7 then 240mg BID on Days 8-10 / 80mg on Day 10 / Simvastatin acid†
Simvastatin
Active inhibitors
Total inhibitors / 2.3
2.5
1.8
1.8 / 2.4
2.1
1.3
1.4
Diltiazem
120mg BID for 10Days / 80mg on Day 10 / Simvastatin acid†
Simvastatin
Active inhibitors
Total inhibitors / 2.7
3.1
2.0
1.7 / 2.7
2.9
1.6
1.5
Amlodipine
10mg QD x 10 Days / 80mg on Day10 / Simvastatin acid†
Simvastatin
Active inhibitors
Total inhibitors / 1.6
1.8
1.3
1.3 / 1.6
1.5
0.9
1.0

*Results based on a chemical assay

†Simvastatin acid refers to the β-hydroxyacid of simvastatin

A single dose of 2g niacin extended-release co-administered with 20 mg simvastatin increased the AUC and Cmax of simvastatin acid by approximately 60% and 84%, respectively, compared to administration of 20 mg simvastatin alone. In this study, the effect of simvastatin on niacin pharmacokinetics was not measured.

The risk of myopathy is increased by high levels of HMG-CoA reductase inhibitory activity in plasma. Potent inhibitors of CYP3A4 can raise the plasma levels of HMG-CoA reductase inhibitory activity and increase the risk of myopathy (see PRECAUTIONS, Myopathy/Rhabdomyolysis;INTERACTIONS WITH OTHER MEDICINES).

Distribution

The mean volume of distribution at steady state following a single 100mg intravenous dose of sitagliptin to healthy subjects is approximately 198litres. The fraction of sitagliptin reversibly bound to plasma proteins is low (38%).

Both simvastatin and βhydroxyacid are bound to human plasma proteins (95%).

Metabolism

Approximately 79% of sitagliptin is excreted unchanged in the urine with metabolism being a minor pathway of elimination. Following a 14C labelled sitagliptin oral dose, approximately 16% of the radioactivity was excreted as metabolites of sitagliptin. Six metabolites were detected at trace levels and are not expected to contribute to the plasma DPP4 inhibitory activity of sitagliptin. In vitro studies indicated that the primary enzyme responsible for the limited metabolism of sitagliptin was CYP3A4, with contribution from CYP2C8.

The major metabolites of simvastatin present in human plasma are βhydroxyacid and four additional active metabolites. Simvastatin and other HMGCoA reductase inhibitors are metabolised by CYP 3A4 (see PRECAUTIONS, Myopathy/Rhabdomyolysis). Although the mechanism is not fully understood, cyclosporin has been shown to increase the AUC of HMG-CoA reductase inhibitors. The increase in AUC for simvastatin acid is presumably due, in part, to inhibition of CYP3A4. Since simvastatin undergoes extensive first-pass extraction in the liver, the availability of simvastatin to the general circulation is low. The availability of β-hydroxyacid to the systemic circulation following an oral dose of simvastatin was estimated using an I.V. reference dose of β-hydroxyacid; the value was found to be less than 5% of the dose.

Excretion

Following administration of an oral 14C labelled sitagliptin dose to healthy subjects, approximately 100% of the administered radioactivity was eliminated in faeces (13%) or urine (87%) within one week of dosing. The apparent terminal t1/2 following a 100mg oral dose of sitagliptin was approximately 12.4hours and renal clearance was approximately 350mL/min. Elimination of sitagliptin occurs primarily via renal excretion and involves active tubular secretion. Sitagliptin is a substrate for human organic anion transporter3 (hOAT3), which may be involved in the renal elimination of sitagliptin. The clinical relevance of hOAT3 in sitagliptin transport has not been established. Sitagliptin is also a substrate of p-glycoprotein, which may also be involved in mediating the renal elimination of sitagliptin. However, cyclosporin, a p-glycoprotein inhibitor, did not reduce the renal clearance of sitagliptin.

Following a 100mg oral dose of 14Clabelled simvastatin in man, 13% of the radioactivity was recovered in urine and 60% in faeces. The latter represents absorbed simvastatin equivalents excreted in bile as well as unabsorbed simvastatin. Less than 0.5% of the dose was recovered in urine as HMGCoA reductase inhibitors. In plasma, the inhibitors account for 14% and 28% (active and total inhibitors) of the AUC of total radioactivity, indicating that the majority of chemical species present were inactive or weak inhibitors. Following an intravenous injection of the β-hydroxyacid metabolite, its half-life averaged 1.9 hours.

Clinical Trials

Sitagliptin

Results from long-term studies of sitagliptin on overall morbidity and mortality outcomes are not available.

There were 2,757 patients with type 2 diabetes randomised in five double-blind, placebo-controlled Phase III clinical studies conducted to evaluate the effects of sitagliptin on glycaemic control as monotherapy and in combination with metformin, pioglitazone, glimepiride, and glimepiride+metformin. Co-morbid diseases were common in the patients studied: 58% had hypertension, 54% had dyslipidaemia, and more than 50% were obese (BMI 30kg/m2). The majority of patients (51.6% to 65.8%) met National Cholesterol Education Program (NCEP) criteria for metabolic syndrome. In these studies, the mean age of patients was 55.0 years, and 62% of patients were white, 18% were Hispanic, 6% were black, 9% were Asian, and 4% were of other racial groups. The studies that support registration in general used the reduction in haemoglobin A1c (HbA1c) as the primary outcome variable. Pre-specified secondary endpoints included fasting plasma glucose (FPG) and 2-hour post-prandial glucose (PPG).

An active (glipizide)-controlled study of 52weeks duration was conducted in 1,172patients with type 2 diabetes who had inadequate glycaemic control on metformin. In patients with type 2 diabetes, treatment with sitagliptin produced statistically significant improvements in HbA1c. Clinically significant improvements in HbA1c were maintained for 52weeks. Treatment with sitagliptin showed suggestions of improvement in measures of beta cell function (see PHARMACOLOGY, Pharmacodynamics).

Add-on Therapy to Metformin

A total of 701patients with type 2 diabetes with inadequate glycaemic control on metformin aloneparticipated in a 24week, randomised, double-blind, placebo-controlled study designed to assess the efficacy of sitagliptin in combination with metformin (Hb A1c 7% to 10%). All patients were started on metformin monotherapy and the dose increased to at least 1,500mg per day. Patients were randomised to the addition of either 100mg of sitagliptin or placebo, administered once daily.

Table2 Glycaemic Parameters and Body Weight at Final Visit (24Week Study)for Sitagliptinin Combination with Metformin† - Primary (HbA1c) and Secondary Outcomes

Sitagliptin 100mg +Metformin / Placebo +
Metformin
HbA1c (%) / N = 453 / N = 224
Baseline (mean) / 7.96 / 8.03
Change from baseline (adjusted mean‡) / -0.67 / -0.02
Difference from placebo + metformin (adjusted mean‡) / -0.65§
Patients (%) achieving HbA1c <7% / 213 (47.0) / 41 (18.3)
FPG (mmol/L) / N = 454 / N = 226
Baseline (mean) / 9.44 / 9.63
Change from baseline (adjusted mean‡) / -0.94 / 0.47
Difference from placebo + metformin (adjusted mean‡) / -1.41§
2-hour PPG (mmol/L) / N = 387 / N = 182
Baseline (mean) / 15.24 / 15.12
Change from baseline (adjusted mean‡) / -3.44 / -0.63
Difference from placebo + metformin (adjusted mean‡) / -2.81§
Body Weight (kg) / N = 399 / N = 169
Baseline (mean) / 86.9 / 87.6
Change from baseline (adjusted mean‡) / -0.7 / -0.6
Difference from placebo + metformin (adjusted mean‡) / -0.1¶

†All Patients Treated Population (an intention-to-treat analysis).

‡Least squares means adjusted for prior antihyperglycaemic therapy and baseline value.

§p<0.001 compared to placebo + metformin.

All Patients as Treated (APaT) population, excluding patients given glycaemic rescue therapy.

¶Not statistically significant (p0.05) compared to placebo + metformin.

In combination with metformin, sitagliptin provided significant improvements in HbA1c (the primary endpoint), FPG, and 2hour PPG compared to placebo with metformin (Table2). A pre-specified secondary endpoint was the number of patients in each group who required therapeutic "rescue" with pioglitazone. Twenty-one of 464 patients (5%) randomised to sitagliptin and 32 of 237 patients (14%) randomised to placebo required pioglitazone "rescue". The improvement in HbA1c compared to placebo was not affected by baseline HbA1c, prior antihyperglycaemic therapy, gender, age, baseline BMI, length of time since diagnosis of diabetes, presence of metabolic syndrome, or standard indices of insulin resistance (HOMAIR) or insulin secretion (HOMAβ). Compared to patients taking placebo, patients taking sitagliptin demonstrated slight decreases in total cholesterol, non-HDL choelsterol and TG. A similar decrease in body weight was observed for both treatment groups.